Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

FOXO1 promotes HIV latency by suppressing ER stress in T cells

Abstract

Quiescence is a hallmark of CD4+ T cells latently infected with human immunodeficiency virus 1 (HIV-1). While reversing this quiescence is an effective approach to reactivate latent HIV from T cells in culture, it can cause deleterious cytokine dysregulation in patients. As a key regulator of T-cell quiescence, FOXO1 promotes latency and suppresses productive HIV infection. We report that, in resting T cells, FOXO1 inhibition impaired autophagy and induced endoplasmic reticulum (ER) stress, thereby activating two associated transcription factors: activating transcription factor 4 (ATF4) and nuclear factor of activated T cells (NFAT). Both factors associate with HIV chromatin and are necessary for HIV reactivation. Indeed, inhibition of protein kinase R-like ER kinase, an ER stress sensor that can mediate the induction of ATF4, and calcineurin, a calcium-dependent regulator of NFAT, synergistically suppressed HIV reactivation induced by FOXO1 inhibition. Thus, our studies uncover a link of FOXO1, ER stress and HIV infection that could be therapeutically exploited to selectively reverse T-cell quiescence and reduce the size of the latent viral reservoir.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: FOXO1 is a specific regulator of HIV latency establishment.
Fig. 2: FOXO1 inhibition reactivates HIV1 from latency.
Fig. 3: FOXO1 inhibition prevents latency establishment and reactivates HIV in primary CD4+ T cells and HIV-infected CD4+ T cells.
Fig. 4: Marked upregulation of ER stress in response to FOXO1 inhibition in primary CD4+ T cells.
Fig. 5: FOXO1 inhibition induces HIV reactivation in the absence of NF-κB recruitment via ATF4 and NFAT.
Fig. 6: Induction of ER stress promotes HIV reactivation.

Similar content being viewed by others

Data availability

The complete array datasets of the RNA-seq data associated with Fig. 4a,b and Extended Data Fig. 4a are available at the Gene Expression Omnibus with the accession number GSE129522. The data that support the findings of the present study are available from the corresponding author upon request. Source data for Figs. 1, 4 and 5 are provided with this paper.

References

  1. Barré-Sinoussi, F., Ross, A. L. & Delfraissy, J.-F. Past, present and future: 30 years of HIV research. Nat. Rev. Microbiol. 11, 877–883 (2013).

    PubMed  Google Scholar 

  2. Archin, N. M., Sung, J. M., Garrido, C., Soriano-Sarabia, N. & Margolis, D. M. Eradicating HIV-1 infection: seeking to clear a persistent pathogen. Nat. Rev. Microbiol. 12, 750–764 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Ruelas, D. S. & Greene, W. C. An integrated overview of HIV-1 latency. Cell 155, 519–529 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Besnard, E. et al. The mTOR complex controls HIV latency. Cell Host Microbe 20, 785–797 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Dahabieh, M. S., Battivelli, E. & Verdin, E. Understanding HIV latency: the road to an HIV cure. Annu. Rev. Med. 66, 407–421 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Spivak, A. M. & Planelles, V. Novel latency reversal agents for HIV-1 cure. Annu. Rev. Med. 69, 421–436 (2018).

    CAS  PubMed  Google Scholar 

  7. Rasmussen, T. A. & Lewin, S. R. Shocking HIV out of hiding: where are we with clinical trials of latency reversing agents? Curr. Opin. HIV AIDS 11, 394–401 (2016).

    CAS  PubMed  Google Scholar 

  8. Manning, B. D. & Toker, A. AKT/PKB signaling: navigating the network. Cell 169, 381–405 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Hedrick, S. M., Michelini, R. H., Doedens, A. L., Goldrath, A. W. & Stone, E. L. FOXO transcription factors throughout T cell biology. Nat. Rev. Immunol. 12, 649–661 (2012).

    CAS  PubMed  Google Scholar 

  10. Webb, A. E., Kundaje, A. & Brunet, A. Characterization of the direct targets of FOXO transcription factors throughout evolution. Aging Cell 15, 673–685 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Accili, D. & Arden, K. C. FoxOs at the crossroads of cellular metabolism, differentiation, and transformation. Cell 117, 421–426 (2004).

    CAS  PubMed  Google Scholar 

  12. Newton, R. H. et al. Maintenance of CD4 T cell fitness through regulation of Foxo1. Nat. Immunol. 19, 838–848 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Gray, S. M., Amezquita, R. A., Guan, T., Kleinstein, S. H. & Kaech, S. M. Polycomb repressive complex 2-mediated chromatin repression guides effector CD8+ T cell terminal differentiation and loss of multipotency. Immunity 46, 596–608 (2017).

  14. Delpoux, A. et al. Continuous activity of Foxo1 is required to prevent anergy and maintain the memory state of CD8+ T cells. J. Exp. Med. 215, 575–594 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Jeng, M. Y. et al. Metabolic reprogramming of human CD8+ memory T cells through loss of SIRT1. J. Exp. Med. 215, 51–62 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. van Grevenynghe, J. et al. Transcription factor FOXO3a controls the persistence of memory CD4+ T cells during HIV infection. Nat. Med. 14, 266–274 (2008).

    PubMed  Google Scholar 

  17. Oteiza, A. & Mechti, N. FoxO4 negatively controls Tat-mediated HIV-1 transcription through the post-transcriptional suppression of Tat encoding mRNA. J. Gen. Virol. 98, 1864–1878 (2017).

    CAS  PubMed  Google Scholar 

  18. Trinité, B. et al. Suppression of Foxo1 activity and down-modulation of CD62L (L-selectin) in HIV-1 infected resting CD4 T cells. PLoS ONE 9, e110719 (2014).

  19. Roux, A. et al. FOXO1 transcription factor plays a key role in T cell—HIV-1 interaction. PLoS Pathog. 15, e1007669 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Battivelli, E. et al. Distinct chromatin functional states correlate with HIV latency reactivation in infected primary CD4+ T cells. eLife 7, e34655 (2018).

  21. Nagashima, T. et al. Discovery of novel forkhead box O1 inhibitors for treating type 2 diabetes: improvement of fasting glycemia in diabetic db/db mice. Mol. Pharmacol. 78, 961–970 (2010).

    CAS  PubMed  Google Scholar 

  22. Shiota, M. et al. Foxo3a suppression of urothelial cancer invasiveness through Twist1, Y-box-binding protein 1, and E-cadherin regulation. Clin. Cancer Res. 16, 5654–5663 (2010).

    CAS  PubMed  Google Scholar 

  23. Tezil, T., Bodur, C., Kutuk, O. & Basaga, H. IKK-β mediates chemoresistance by sequestering FOXO3: a critical factor for cell survival and death. Cell. Signal. 24, 1361–1368 (2012).

    CAS  PubMed  Google Scholar 

  24. Jordan, A., Bisgrove, D. & Verdin, E. HIV reproducibly establishes a latent infection after acute infection of T cells in vitro. EMBO J. 22, 1868–1877 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Laird, G. M. et al. Ex vivo analysis identifies effective HIV-1 latency-reversing drug combinations. J. Clin. Invest. 125, 1901–1912 (2015).

    PubMed  PubMed Central  Google Scholar 

  26. Bliss, C. I. The toxicity of poisons applied jointly. Ann. Appl. Biol. 26, 585–615 (1939).

    CAS  Google Scholar 

  27. Lassen, K. G., Hebbeler, A. M., Bhattacharyya, D., Lobritz, M. A. & Greene, W. C. A flexible model of HIV-1 latency permitting evaluation of many primary CD4 T-cell reservoirs. PLoS ONE 7, e30176 (2012).

  28. Reuse, S. et al. Synergistic activation of HIV-1 expression by deacetylase inhibitors and prostratin: Implications for treatment of latent infection. PLoS ONE 4, e6093 (2009).

  29. Zhao, Y. et al. Cytosolic FoxO1 is essential for the induction of autophagy and tumour suppressor activity. Nat. Cell Biol. 12, 665–675 (2010).

    CAS  PubMed  Google Scholar 

  30. Webb, A. E. & Brunnet, A. FOXO transcription factors: key regulators of cellular quality control. Trends Biochem. Sci. 39, 159–169 (2015).

    Google Scholar 

  31. Ron, D. & Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell Biol. 8, 519–529 (2007).

    CAS  PubMed  Google Scholar 

  32. Pakos-Zebrucka, K. et al. The integrated stress response. EMBO Rep. 17, 1374–1395 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Kilberg, M. S., Shan, J. & Su, N. ATF4-dependent transcription mediates signaling of amino acid limitation. Trends Endocrinol. Metab. 20, 436–443 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Castilho, B. A. et al. Keeping the eIF2 alpha kinase Gcn2 in check. Biochim. Biophys. Acta 1843, 1948–1968 (2014).

    CAS  PubMed  Google Scholar 

  35. Carreras-Sureda, A., Pihán, P. & Hetz, C. Calcium signaling at the endoplasmic reticulum: fine-tuning stress responses. Cell Calcium 70, 24–31 (2018).

    CAS  PubMed  Google Scholar 

  36. Deniaud, A. et al. Endoplasmic reticulum stress induces calcium-dependent permeability transition, mitochondrial outer membrane permeabilization and apoptosis. Oncogene 27, 285–299 (2008).

    CAS  PubMed  Google Scholar 

  37. Jiang, G. et al. HIV exploits antiviral host innate GCN2-ATF4 signaling for establishing viral replication early in infection. mBio 8, 1518–1534 (2018).

    Google Scholar 

  38. Reddy, T. R., Tang, H., Li, X. & Wong-Staal, F. Functional interaction of the HTLV-1 transactivator Tax with activating transcription factor-4 (ATF4). Oncogene 14, 2785–2792 (1997).

    CAS  PubMed  Google Scholar 

  39. Flanagan, W. F., Corthesy, B., Bram, R. J. & Crabtree, G. R. Nuclear association of a T-cell transcription factor blocked by FK-506 and cyclosporin A. Nature 352, 803–807 (1991).

    CAS  PubMed  Google Scholar 

  40. Clipstone, N. A. & Crabtree, G. R. Identification of calcineurin as a key signalling enzyme in T lymphocyte activation. Nature 356, 695–697 (1992).

    Google Scholar 

  41. Schütze, S., Wiegmann, K., Machleidt, T. & Krönke, M. TNF-induced activation of NF-κB. Immunobiology 193, 193–203 (1995).

    PubMed  Google Scholar 

  42. Kinoshita, S., Chen, B. K., Kaneshima, H. & Nolan, G. P. Host control of HIV-1 parasitism in T cells by the nuclear factor of activated T cells. Cell 95, 595–604 (1998).

    CAS  PubMed  Google Scholar 

  43. So, J.-S. Roles of endoplasmic reticulum stress in immune responses. Mol. Cell 41, 705–716 (2018).

    CAS  Google Scholar 

  44. Walter, P. & Ron, D. The unfolded protein response: from stress pathway to homeostatic regulation. Mol. Biotechnol. 34, 279–290 (2006).

    Google Scholar 

  45. Harding, H. P. et al. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol. Cell 6, 1099–1108 (2000).

    CAS  PubMed  Google Scholar 

  46. Jammi, N. V., Whitby, L. R. & Beal, P. A. Small molecule inhibitors of the RNA-dependent protein kinase. Biochem. Biophys. Res. Commun. 308, 50–57 (2003).

    CAS  PubMed  Google Scholar 

  47. Axten, M. et al. Discovery of GSK2656157: an optimized PERK inhibitor selected for preclinical development. J. Med. Chem. 4, 964–968 (2013).

    CAS  Google Scholar 

  48. Brazeau, J.-F. & Rosse, G. Triazolo[4,5-d]pyrimidine derivatives as inhibitors of GCN2. ACS Med. Chem. Lett. 5, 282–283 (2014).

    PubMed  PubMed Central  Google Scholar 

  49. Cron, R. Q. et al. NFAT1 enhances HIV-1 gene expression in primary human CD4 T cells. Clin. Immunol. 94, 179–191 (2000).

    CAS  PubMed  Google Scholar 

  50. Corazzari, M. et al. Targeting homeostatic mechanisms of endoplasmic reticulum stress to increase susceptibility of cancer cells to fenretinide-induced apoptosis: the role of stress proteins ERdj5 and ERp57. Br. J. Cancer 96, 1062–1071 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Hail, N., Kim, H. J. & Lotan, R. Mechanisms of fenretinide-induced apoptosis. Apoptosis 11, 1677–1694 (2006).

    CAS  PubMed  Google Scholar 

  52. Thaxton, J. E. et al. Modulation of endoplasmic reticulum stress controls CD4+ T-cell activation and antitumor function. Cancer Immunol. Res. 5, 666–675 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Kops, G. J. P. L. et al. Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature 419, 316–321 (2002).

    CAS  PubMed  Google Scholar 

  54. Kode, A. et al. FoxO1 protein cooperates with ATF4 protein in osteoblasts to control glucose homeostasis. J. Biol. Chem. 287, 8757–8768 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Sundaresan, N. R. et al. Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J. Clin. Invest. 119, 2758–2771 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Ni, Y. G. et al. Foxo transcription factors blunt cardiac hypertrophy by inhibiting calcineurin signaling. Circulation 114, 1159–1168 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. De Leo, A., Chen, H. S., Hu, C. C. A. & Lieberman, P. M. Deregulation of KSHV latency conformation by ER-stress and caspase-dependent RAD21-cleavage. PLoS Pathog. 13, e1006596 (2017).

    PubMed  PubMed Central  Google Scholar 

  58. Chan, S. & Egan, P. A. Hepatitis C virus envelope proteins regulate CHOP via induction of the unfolded protein response. FASEB J. 19, 1510–1512 (2005).

    CAS  PubMed  Google Scholar 

  59. Medigeshi, G. R. et al. West Nile virus infection activates the unfolded protein response, leading to CHOP induction and apoptosis. J. Virol. 81, 10849–10860 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Bitko, V. & Barik, S. An endoplasmic reticulum-specifc stress-activated caspase (caspase-12) is implicated in the apoptosis of A549 epithelial cells by respiratory syncytial virus. J. Cell. Biochem. 80, 441–454 (2001).

    CAS  PubMed  Google Scholar 

  61. Su, H., Liao, C. & Lin, Y. Japanese encephalitis virus infection initiates endoplasmic reticulum stress and an unfolded protein response. J. Virol. 76, 4162–4171 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Isler, J. A., Skalet, A. H. & Alwine, J. C. Human cytomegalovirus infection activates and regulates the unfolded protein response. J. Virol. 79, 6890–6899 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Fraser, J. E., Wang, C., Chan, K. W. K., Vasudevan, S. G. & Jans, D. A. Novel dengue virus inhibitor 4-HPR activates ATF4 independent of protein kinase R like endoplasmic reticulum kinase and elevates levels of eIF2a phosphorylation in virus infected cells. Antiviral Res. 130, 1–6 (2016).

    CAS  PubMed  Google Scholar 

  64. Liao, Y. et al. Upregulation of CHOP/GADD153 during coronavirus infectious bronchitis virus infection modulates apoptosis by restricting activation of the extracellular signal-regulated kinase pathway. J. Virol. 87, 8124–8134 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Borsa, M. et al. HIV infection and antiretroviral therapy lead to unfolded protein response activation retroviruses. Virol. J. 12, 77 (2015).

    PubMed  PubMed Central  Google Scholar 

  66. Caselli, E., Benedetti, S., Gentili, V., Grigolato, J. & Di Luca, D. Short communication: activating transcription factor 4 (ATF4) promotes HIV type 1 activation. AIDS Res. Hum. Retroviruses 28, 907–912 (2012).

    CAS  PubMed  Google Scholar 

  67. Lee, S. D. et al. Understanding of the functional role(s) of the activating transcription factor 4(ATF4) in HIV regulation and production. BMB Rep. 51, 388–393 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Fan, Y. & He, J. J. HIV-1 tat induces unfolded protein response and endoplasmic reticulum stress in astrocytes and causes neurotoxicity through glial fibrillary acidic protein (GFAP) activation and aggregation. J. Biol. Chem. 291, 22819–22829 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Jiang, S., Zhang, E., Zhang, R. & Li, X. Altered activity patterns of transcription factors induced by endoplasmic reticulum stress. BMC Biochem. 17, 8 (2016).

    PubMed  PubMed Central  Google Scholar 

  70. Rizzuto, R., Hendershot, L., Meldolesi, J. & Lievremont, J.-P. BiP, a major chaperone protein of the endoplasmic reticulum lumen, plays a direct and important role in the storage of the rapidly exchanging pool of Ca2+. J. Biol. Chem. 272, 30873–30879 (1997).

    PubMed  Google Scholar 

  71. Vattem, K. M. & Wek, R. C. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proc. Natl Acad. Sci. USA 101, 11269–11274 (2004).

    CAS  PubMed  Google Scholar 

  72. Mammucari, C. et al. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab. 6, 458–471 (2007).

    CAS  PubMed  Google Scholar 

  73. Juhász, G. et al. Gene expression profiling identifies FKBP39 as an inhibitor of autophagy in larval Drosophila fat body. Cell Death Differ. 14, 1181–1190 (2007).

    PubMed  PubMed Central  Google Scholar 

  74. Zhang, W. et al. ER stress potentiates insulin resistance through PERK-mediated FOXO phosphorylation. Genes Dev. 27, 441–449 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Zou, P. et al. Targeting FoxO1 with AS1842856 suppresses adipogenesis. Cell Cycle 13, 3759–3767 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. CerRx, Inc. Trial of intravenous fenretinide emulsion for patients with relapsed/refractory peripheral T-cell lymphomas. Primary ID: NCT02495415, Secondary ID: NCI-2015-01195, FEN T-14. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02495415 (2019).

  77. Tsuji, G., Okiyama, N., Villarroel, V. A. & Katz, S. I. Histone deacetylase 6 inhibition impairs effector CD8 T-cell functions during skin inflammation. J. Allergy Clin. Immunol. 135, 1228–1239 (2015).

    CAS  PubMed  Google Scholar 

  78. Chan, J. K., Bhattacharyya, D., Lassen, K. G., Ruelas, D. & Greene, W. C. Calcium/calcineurin synergizes with prostratin to promote NF-κB dependent activation of latent HIV. PLoS ONE 8, e77749 (2013).

  79. Gilbert, L. A. et al. Resource genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Geng, X., Doitsh, G., Yang, Z., Galloway, N. L. K. & Greene, W. C. Efficient delivery of lentiviral vectors into resting human CD4 T cells. Gene Ther. 21, 444–449 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Goadsby, P. J., Kurth, T. & Pressman, A. Fluorescence-linked antigen quantification (FLAQ) assay for fast quantification of HIV-1 p24Gag. Bio Protoc. 35, e1366 (2016).

    Google Scholar 

  82. Battivelli, E. & Verdin, E. HIVGKO: a tool to assess HIV-1 latency reversal agents in human primary CD4+ T cells. Bio Protoc. 8, e3050 (2018).

    CAS  Google Scholar 

  83. Horlbeck, M. A. et al. Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation. eLife 9, e19760 (2016).

    Google Scholar 

  84. Ianevski, A., He, L., Aittokallio, T. & Tang, J. SynergyFinder: a web application for analyzing drug combination dose–response matrix data. Bioinformatics 33, 2413–2415 (2017).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all members of the Ott, Verdin and Pillai laboratories for helpful discussions, reagents and expertise. We thank D. Lacuadra and A. Patel for assistance, M. Cavrois, N. Raman and the Gladstone Flow Cytometry Core for assistance with FACS, N. Carli, J. McGuire and the Gladstone Genomics Core for assistance with RNA-seq, J. Carroll for graphics, K. Claiborn and B. Mensh for editorial assistance, and V. Fonseca and L. Weiser for administrative assistance. This work was supported by the National Institutes of Health/National Institute of Allergy and Infectious Diseases (NIH/NIAID, grant nos. DP1DA038043 and R37AI083139 to M.O.), the NIH/National Institute on Drug Abuse (grant nos. R01DA041742 and R01AI117864 to E.V.), the NIH/National Institute of General Medical Sciences (grant no. R01GM117901 to S.K.P.) and the NIH (grant no. P30 AI027763 to Flow Cytometry Core). Research was also supported as part of the amfAR Institute for HIV Cure Research, with funding from amfAR (grant no. 109301). D.B. was also funded by the Gilead HIV Cure Mentored Scientist Award from the amfAR Institute for HIV Cure Research at the UCSF AIDS Research Institute.

Author information

Authors and Affiliations

Authors

Contributions

A.V.-G. and M.O. designed and guided the study. A.V.-G., I.C. and R.P. designed, performed and analysed most biochemical experiments. E.B. helped design the project. D.B. performed ChIP assays. E.B. helped with primary cell experiments. T.T. performed CETSAs. K.K., K.R., P.A.H., M.W., I.H. and A.C. performed some biochemical experiments. S.D. provided resources. S.P., E.V. and M.O. provided the use of their laboratories and helped with data interpretation. The manuscript was written by A.V.-G. and M.O.

Corresponding author

Correspondence to Melanie Ott.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Extended Data Fig 1.

a, Jurkat-derived NH7(Cas9) cells were treated with increasing concentrations of the FOXO1 inhibitor AS1842856 just after HIVGKO infection. After 3-4 days, the percentage of latent and productively infected cells were quantified by FACS. In left panel, percentage of productively or latently cells relative to the total infection rate (bars) and total cell viability (blue dotted line) of a representative experiment. In the right panel, ratios of productive versus latent populations relative to the total infection rate upon increasing concentrations of AS1842856 treatments. Data are represented as mean ± SD of triplicate values, representative of n = 3 independent experiments. b, Representative western blots of CETSA assays in FOXO1 and FOXO3 in the presence or absence of 100 nM AS1842856. c, FOXO1 and FOXO3 CETSA-melting curves upon the presence or absence of AS1842856 100 nM in K562(Cas9) cells. Band intensities obtained from western blot analysis were normalized to the highest western blot signal which has been set to 100 %. Relative FOXO-band intensities were plotted against corresponding incubation temperatures and a nonlinear least-squares regression fit was applied. Data represent the mean ± SD of n = 3 individual experiments. d, Cell viability of CETSA assay in FOXO1 experiments assessed by Trypan Blue exclusion. Data represent the mean ± SD of three individual experiments. e, FOXO1, FOXO3 and FOXO4 gene expression in KD-1A-FOXO1 K562(Cas9) cell line analyzed by RT-qPCR, normalized to RPL13A mRNA. Data represent mean ± SD of n = 3 individual experiments, except for FOXO1 (n = 4). f, Cell growth analysis of WT, FOXO1, FOXO3 and FOXO4 knockdowns K562(Cas9) cell lines. Data are represented as mean ± SD of triplicate values. g-i, Percentage of productive or latent cells relative to the total infection rate and cell viability upon increasing concentrations of AS1842856 treatment in the WT K562(Cas9) (g) or in the FOXO1-knockdown cell lines KD-1A (h) and KD-1B (i). Data are represented as mean ± SD of n = 2 independent experiments.

Extended Data Fig. 2 Extended Data Fig 2.

a, J-Lat cell lines 5A8, 6.3, 11.1 and 15.4 were treated with increasing concentrations of AS1842856 for 24, 48 and 72 h and HIV-GFP reactivation was analyzed by flow cytometry. HIV-GFP reactivation is reported as a Mean Intensity Fluorescence (MFI) of GFP-expressing cells. Data represent mean ± SD of n ≥ 3 independent experiments. 10 ng/mL TNFα was used as control. b, J-Lat cell lines 5A8, 6.3, 11.1 and 15.4 were treated for 72 h with increasing concentrations of both AS1842856 (Y-axis) and TNFα (X-axis) alone or in combination and analyzed by FACS. HIV-GFP reactivation is reported as a percentage of GFP-expressing cells (% GFP + cells) or c, Mean Intensity Fluorescence (MFI). Data represent mean of n = 3 independent experiments.

Extended Data Fig. 3 Extended Data Fig 3.

a, HIV reactivation was measured by luciferase activity and cell viability by flow cytometry assessed in CD4+ T cells purified from blood of healthy donors and infected with HIVNL4-3 Luciferase and letting them rest for 6 days before reactivation was induced with 10 µg/mL PHA + 100 U/mL IL-2 and 10 µg/mL αCD3 + 1 µg/mL αCD28 for 72 h, in the presence of raltegravir (30 μM). Data represent average ± SD of n ≥ 3 independent experiments. b, The cell surface CD69 and CD25 T cell activation markers were measured by FACS in CD4+ T cells upon AS1842856 treatment for 24, 48 and 72 h. 10 µg/mL αCD3 and 1 µg/mL αCD28 was used as control. Data is shown as mean of percentage of positive cells and as mean ± SD of n = 2 biological replicates.

Extended Data Fig. 4 Extended Data Fig 4.

a, Venn diagrams (top panels) comparing the up- and down-regulated genes from a recently published Affimmetrix microarray19 and RNA-Seq data presented here (Vallejo-Gracia et al.). GO Enrichment Analysis (Biological Processes and Cellular Components) from overlapped dysregulated genes from the two datasets. b, Confirmation of selected up- or down-regulated genes and pathways after 72 h AS1842856 treatment in CD4+ T cells of HIV-infected patients on antiretroviral-therapy with undetectable viral load by RT-qPCR, normalized to RPL13A mRNA. Data represent mean ± SD of n = 10 individual donors. c, The thresholded Mander’s correlation coefficients were determined and P value was calculated by unpaired Student’s t test. n = 90 cells per condition. Data are represented as mean ± SD.

Extended Data Fig. 5 Extended Data Fig 5.

Representative plots of intracellular calcium-flux kinetics in K562(Cas9) and Jurkat cell lines in the presence or absence of AS1842856 (100 nM). Cells were stained with a membrane permeable calcium sensor dye in PBS and stimulated by adding Ionomycin after 30 seconds resulting in an increase of fluorescence indicating a calcium mobilization from the ER. Representative experiments of n = 3 independent experiments.

Extended Data Fig. 6 Extended Data Fig 6.

a, J-Lat cell line A58 was treated with increasing concentrations of GCN2i (A-92) in combination with 1,000 nM AS1842856 (72 h) or 10 ng/mL TNFα (24 h). In the upper panel, HIV-GFP reactivation was analyzed by FACS and relativized to the control. In the lower panel, histogram plots of percent live cells for each drug treatment are shown. Data are represented by mean ± SD of n = 3 different experiments. b, Same experiment as in Extended Data Fig. 6a but treating cells with increasing concentrations of PKRi (Imidazolo-oxindole PKR inhibitor C16). Data are represented by mean ± SD of three different experiments. c, Same experiment as in Extended Data Fig. 6a but treating cells with increasing concentrations of PERKi (GSK2656157 / PERK inhibitor II) (top panels) or the highly specific PERK inhibitor (AMG PERK 44) (lower panels). Histogram plots of percent live cells for each drug treatment are shown. Data represent mean ± SD of n = 3 independent experiments. d, Same experiment as in Extended Data Fig. 6a, but cells were treated with increasing concentrations of IRE1αi (MKC8866). Data are represented by mean ± SD of n = 3 different experiments. e, J-Lat cell line A58 was treated with increasing concentrations of IRE1αi (MKC8866) in combination with 1 µM Thapsigargin (6 h). Data are represented by mean ± SD of n = 3 different experiments. f, Same experiment as in Extended Data Fig. 6a but treating cells with increasing concentrations of CsA (Cyclosporin A) (left panels) or the combined concentrations of PERKi and Cyclosporin A (right panels). Histogram plots of percent live cells for each drug treatment are shown. Data represent mean ± SD of n ≥ 3 independent experiments. g, J-Lat cell line A58 was treated with increasing concentrations of Thapsigargin (0.01, 0.1, 1 µM), Brefeldin A (0.01, 0.1, 1 µg/mL) and Fenretinide (0.5, 2, 5 µM) for 24, 48 and 72 h (bottom right). Histogram plots of percent live cells for each drug treatment are shown. Data represent mean ± SD of n = 3 independent experiments. h, J-Lat cell line A58 was treated with increasing concentrations of Ionomycin (0.01, 0.1, 0.5, 1 µM) for 24, 48 and 72 h and HIV-GFP reactivation (upper panel) and cell viability (lower panel) were analyzed by FACS. Data represent mean ± SD of n = 3 independent experiments. i, J-Lat cell line 5A8 was treated for 72 h with increasing concentrations of both Fenretinide (Y-axis) and Ionomycin (X-axis) alone or in combination and analyzed by FACS. HIV-GFP reactivation is reported as a percentage of GFP-expressing cells (% GFP + cells) (upper panel) and viability was measured by FACS (bottom panel). Data represent average of n = 3 independent experiments.

Supplementary information

Reporting Summary

Peer Review Information

Reviewer reports and authors’ response from the peer review of this Article at Nature Microbiology.

Source data

Source Data Fig. 1

Raw blots for Fig. 1c.

Source Data Fig. 4

Raw blots for Fig. 4d.

Source Data Fig. 5

Raw blots for Fig. 5b.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vallejo-Gracia, A., Chen, I.P., Perrone, R. et al. FOXO1 promotes HIV latency by suppressing ER stress in T cells. Nat Microbiol 5, 1144–1157 (2020). https://doi.org/10.1038/s41564-020-0742-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-020-0742-9

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing